Chronic cyclodextrin treatment of murine Niemann-Pick C disease ameliorates neuronal cholesterol and glycosphingolipid storage and disease progression

Cristin D Davidson, Nafeeza F Ali, Matthew C Micsenyi, Gloria Stephney, Sophie Renault, Kostantin Dobrenis, Daniel S Ory, Marie T Vanier, Steven U Walkley, Cristin D Davidson, Nafeeza F Ali, Matthew C Micsenyi, Gloria Stephney, Sophie Renault, Kostantin Dobrenis, Daniel S Ory, Marie T Vanier, Steven U Walkley

Abstract

Background: Niemann-Pick type C (NPC) disease is a fatal neurodegenerative disorder caused most commonly by a defect in the NPC1 protein and characterized by widespread intracellular accumulation of unesterified cholesterol and glycosphingolipids (GSLs). While current treatment therapies are limited, a few drugs tested in Npc1(-/-) mice have shown partial benefit. During a combination treatment trial using two such compounds, N-butyldeoxynojirimycin (NB-DNJ) and allopregnanolone, we noted increased lifespan for Npc1(-/-) mice receiving only 2-hydroxypropyl-beta-cyclodextrin (CD), the vehicle for allopregnanolone. This finding suggested that administration of CD alone, but with greater frequency, might provide additional benefit.

Methodology/principal findings: Administration of CD to Npc1(-/-) mice beginning at either P7 or P21 and continuing every other day delayed clinical onset, reduced intraneuronal cholesterol and GSL storage as well as free sphingosine accumulation, reduced markers of neurodegeneration, and led to longer survival than any previous treatment regime. We reasoned that other lysosomal diseases characterized by cholesterol and GSL accumulation, including NPC disease due to NPC2 deficiency, GM1 gangliosidosis and mucopolysaccharidosis (MPS) type IIIA, might likewise benefit from CD treatment. Treated Npc2(-/-) mice showed benefits similar to NPC1 disease, however, mice with GM1 gangliosidosis or MPS IIIA failed to show reduction in storage.

Conclusions/significance: Treatment with CD delayed clinical disease onset, reduced intraneuronal storage and secondary markers of neurodegeneration, and significantly increased lifespan of both Npc1(-/-) and Npc2(-/-) mice. In contrast, CD failed to ameliorate cholesterol or glycosphingolipid storage in GM1 gangliosidosis and MPS IIIA disease. Understanding the mechanism(s) by which CD leads to reduced neuronal storage may provide important new opportunities for treatment of NPC and related neurodegenerative diseases characterized by cholesterol dyshomeostasis.

Conflict of interest statement

Competing Interests: SUW is an author on a patent for the use of miglustat for the treatment of Niemann-Pick C disease. MTV has been a paid and non-paid consultant for Actelion, which markets miglustat as treatment for Niemann-Pick C disease.

Figures

Figure 1. Combination treatment using N B-DNJ…
Figure 1. Combination treatment using NB-DNJ and allopregnanolone/CD in Npc1−/− mice.
(A) Average weight over time for each treatment group shown for males and females separately. (B) Survival of each treatment group. Median survival of Npc1−/− mice: no treatment, 79 days; CD (weekly), 118 days; Miglustat (daily), 123 days; Allopregnanolone/CD (weekly), 146 days; Combination therapy (Miglustat + Allopregnanolone/CD), 152 days.
Figure 2. Cholesterol and ganglioside immunohistochemistry (IHC)…
Figure 2. Cholesterol and ganglioside immunohistochemistry (IHC) of Npc1−/− and WT mice in the combination treatment study.
(A) Filipin labeling of unesterified cholesterol (seen as white areas in image) in the neocortex of age-matched untreated and treated Npc1−/− and WT mice (all mice between 75 and 81 days of age) revealed less cholesterol accumulation of treated Npc1−/− mice (second, third, and fourth panels) when compared to control Npc1−/− mice (fifth panel). WT mice do not exhibit cholesterol accumulation (first panel). Each panel here and in (B) and (C) shows layers II (top) through VI (bottom) of the cerebral cortex. (B) IHC of GM2 ganglioside (visualized as brown punctae within cells) was also characterized by reduced GM2 storage in all treated Npc1−/− mice. (C) IHC of GM3 ganglioside (again seen as brown punctae within cells) showed results similar to GM2. (D) Treated Npc1−/− mice had more remaining Purkinje cells (brown areas in cerebellar images) than did untreated Npc1−/− mice; however, treated mice still had Purkinje cell loss when compared to WT mice. Anti-calbindin antibody labels Purkinje cell bodies and dendritic arbors, while the Nissl counterstain (purple) labels all neuronal cell bodies. Images taken at 20X (A), 10X (B, C), and 2X (D); scale bars 20 µm (A), 50 µm (B, C), and 400 µm (D).
Figure 3. Short term (2 week) CD…
Figure 3. Short term (2 week) CD study in Npc1−/− mice.
(A) Filipin labeling of unesterified cholesterol in the neocortex of untreated and CD-treated Npc1−/− mice revealed dramatically less cholesterol accumulation in CD-treated mice at 22 days of age. Mice were administered SC injections of CD every other day for 2 weeks starting at P7. (B) IHC of untreated and CD-treated Npc1−/− mice also revealed less GM2 storage present in CD-treated mice (similar finding for GM3, not shown). (C) Biochemical analysis of ganglioside levels further corroborated the reduction in GM2 and GM3 seen with IHC analysis. (D) Ultrastructural analysis of neocortical neurons in Npc1−/− untreated and CD-treated mice showed remarkably normal neuronal morphology in CD-treated mice. (E) Filipin labeling of unesterified cholesterol in the cerebellum of untreated and CD-treated Npc1−/− mice indicated little to no cholesterol accumulation present within Purkinje cells of CD-treated mice; cerebellar layers: molecular cell layer (MCL), Purkinje cell layer (PCL), and granular cell layer (GCL). (F) Western blot analysis of LC3-II, an autophagosome marker, revealed less LC3-II present in CD-treated Npc1−/− as compared to untreated Npc1−/− mice. Images taken at 20X (A, E) and 10X (B); scale bars 20 µm (A, E), 50 µm (B), 1 µm (D).
Figure 4. Chronic allopregnanolone and chronic CD…
Figure 4. Chronic allopregnanolone and chronic CD treatment studies in Npc1−/− mice.
(A) Survival of untreated and allopregnanolone-treated Npc1−/− mice using different vehicles for allopregnanolone. Median survival of Npc1−/− mice: DMSO, 84 days; Allo/DMSO, 83 days; Corn Oil, 81 days; Allo/Corn Oil, 88 days; 5% CD, 114 days; Allo/5% CD, 121 days. (B) Average weight over time for untreated and chronically CD-treated Npc1−/− and WT mice. Weights of untreated and CD-treated WTs were averaged for each gender as there was no significant difference between treatments (p<0.8125). (C) Survival of untreated and CD-treated Npc1−/− and WT mice showing effects of different start times. Median survival of Npc1−/− mice: no treatment, 83 days; CD (every other day, start at P7), 185 days; CD (every other day, start at P21), 149 days. Treatment initiated at P7 appeared more efficacious, although lifespan was not significantly longer when compared to treatment initiated at P21 (p<0.1870).
Figure 5. Chronic CD treatment study in…
Figure 5. Chronic CD treatment study in Npc1−/− mice.
(A) Filipin labeling of unesterified cholesterol in the neocortex of untreated (end-stage, 78 days old) and CD-treated (start at P21; end-stage, 197 days old) Npc1−/− mice showed reduced cholesterol accumulation in a CD-treated mouse. (B) Higher magnification of neocortex in same CD-treated Npc1−/− animal as previous panel, showed presence of neurons with cholesterol accumulation while neighboring cells lacked this storage. (C) Confocal microscopy further revealed that gangliosides and cholesterol appeared to always co-sequester within neurons in an untreated Npc1−/− mouse (end-stage, 78 days old; upper panel). However, some neocortical neurons in a CD-treated Npc1−/− mouse (start at P7; end-stage, 182 days old; lower panel) had little to no detectable cholesterol accumulation, yet still exhibited ganglioside storage. Cholesterol (red, visualized with BC Theta), GM2 (blue), and GM3 (green); n denotes nucleus of single neuron shown in each image. (D) IHC of untreated and CD-treated Npc1−/− mice (same mice as A), revealed less GM2 storage (also GM3, not shown) in the neocortex of a CD-treated mouse. (E) Biochemical analysis of ganglioside levels further corroborated the reduction in GM2 and GM3 seen with IHC. Data from WT and untreated mutant mice represent mean ± SD. (F) Ultrastructural analysis of neocortical neurons in untreated and CD-treated Npc1−/− mice (same mice as C) revealed presence of PCBs in both groups, but CD-treated mice appeared to have fewer of these storage bodies. (G) Western blot analysis of LC3-II in 85 day old untreated and CD-treated (start at P21) Npc1−/− and WT mice revealed a reduction in LC3-II levels in the CD-treated Npc1−/− mouse. Images taken at 20X (A), 40X (B), 63X (C), and 10X (D); scale bars 20 µm (A, B), 2 µm (C), 50 µm (D), 1 µm (F).
Figure 6. Chronic CD treatment study in…
Figure 6. Chronic CD treatment study in Npc2−/− mice.
(A) Average weight over time for untreated and CD-treated Npc2−/− and WT mice. Weights of untreated and CD-treated WTs were averaged for each gender as there was no significant difference between treatments (p<0.8125). (B) Survival of untreated and CD-treated Npc2−/− and WT mice. Median survival of Npc2−/− mice: no treatment, 144 days; CD (every other day), 248 days. CD-treated Npc2−/− mice lived significantly longer than untreated Npc2−/− mice (p<0.0108). (C) Filipin labeling of unesterified cholesterol in neocortex of untreated (end-stage, 145 days old) and CD-treated (start at P9; 150 days old) Npc2−/− mice revealed less cholesterol accumulation in the CD-treated mouse. (D) IHC of the same untreated and CD-treated Npc2−/− mice showed reduced GM2 labeling in the neocortex of the CD-treated mouse (also for GM3, not shown). (E) Biochemical analysis of ganglioside levels confirmed the reduction in GM2 and GM3 storage seen with IHC. Data from WT and mutant mice represent mean ± SD. Images taken at 20X (C) and 10X (D); scale bars 20 µm (C) and 50 µm (D).
Figure 7. Free sphingosine concentrations in NPC…
Figure 7. Free sphingosine concentrations in NPC disease following CD treatment.
(A) Biochemical analysis of sphingosine in brain of untreated and CD-treated Npc1−/− and WT mice revealed that chronic CD treatment reduced sphingosine concentrations in affected mice, even when treatment was initiated post-weaning. Npc2−/− mice exhibited a similar trend, although the effect was less pronounced. (B) Biochemical analysis of liver from untreated and CD-treated Npc1−/− and Npc2−/− mice showed that with chronic CD treatment, sphingosine accumulation was reduced to levels near those found in WT.
Figure 8. Chronic CD treatment study in…
Figure 8. Chronic CD treatment study in GM1 and MPS IIIA mice.
(A, B) Filipin labeling of unesterified cholesterol (A) and IHC of GM1 ganglioside (B) in neocortex of untreated (138 days old) and CD-treated (start at P21; 138 days old) mice with GM1 gangliosidosis. Brown punctae indicate GM1 accumulation within cortical neurons. No differences were observed in either filipin or GM1 labeling between CD-treated and untreated GM1 mice. (C, D) Filipin labeling of unesterified cholesterol (C) and IHC of GM2 ganglioside (D) in neocortex of untreated (140 days old) and CD-treated (start at P30; 140 days old) MPS IIIA mice. As with GM1 mice, no differences were observed between CD-treated and untreated MPS IIIA mice. (E) No reductions in GM1, Asialo-GM1, GM2, or GM3 ganglioside levels were seen in cerebral homogenates of CD-treated GM1 and MPS IIIA mice compared to untreated mice as evidenced by the thin-layer chromatography plate. Images taken at 20X (A, C) and 10X (B, D); scale bars 20 µm (C, also applicable to A) and 50 µm (D, also applicable to B).

References

    1. Pentchev PG, Comly ME, Kruth HS, Vanier MT, Wenger DA, et al. A defect in cholesterol esterification in Niemann-Pick disease (type C) patients. PNAS USA. 1985;82:8247–8251.
    1. Patterson MC, Vanier MT, Suzuki K, Morris JA, Carstea E, et al. Niemann-Pick disease type C: a lipid trafficking disorder. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. New York: McGraw-Hill Medical Publishing Division; 2001. pp. 3611–3633. The Metabolic & Molecular Bases of Inherited Disease, vol. 3, 8th edition.
    1. Walkley SU, Suzuki K. Consequences of NPC1 and NPC2 loss of function in mammalian neurons. Biochim Biophys Acta. 2004;1685:48–62.
    1. Sarna JR, Larouche M, Marzban H, Sillitoe RV, Rancourt DE, et al. Patterned Purkinje cell degeneration in mouse models of Niemann-Pick type C disease. J Comp Neurol. 2003;456:279–291.
    1. Ohgami N, Ko DC, Thomas M, Scott MP, Chang CC, et al. Binding between the Niemann-Pick C1 protein and a photactivatable cholesterol analog requires a functional sterol-sensing domain. PNAS USA. 2004;101:12473–12478.
    1. Naureckiene S, Sleat DE, Lackland H, Fensom A, Vanier MT, et al. Identification of HE1 as the second gene of Niemann-Pick C disease. Science. 2000;290:2298–2301.
    1. Ko DC, Binkley J, Sidow A, Scott MP. The integrity of a cholesterol-binding pocket in Niemann-Pick C2 protein is necessary to control lysosome cholesterol levels. PNAS USA. 2003;100:2518–2525.
    1. Infante RE, Abi-Mosleh L, Radhakrishnan A, Dale JD, Brown MS, et al. Purified NPC1 protein. I. Binding of cholesterol and oxysterols to a 1278-amino acid membrane protein. J Biol Chem. 2008;283:1052–1063.
    1. Infante RE, Wang ML, Radhakrishnan A, Kwon HJ, Brown MS, et al. NPC2 facilitates bidirectional transfer of cholesterol between NPC1 and lipid bilayers, a step in cholesterol egress from lysosomes. PNAS USA. 2008;105:15287–15292.
    1. Sleat DE, Wiseman JA, El-Banna M, Price SM, Verot L, et al. Genetic evidence for nonredundant functional cooperativity between NPC1 and NPC2 in lipid transport. PNAS USA. 2004;101:5886–5891.
    1. Liu Y, Wu YP, Wada R, Neufeld EB, Mullin KA, et al. Alleviation of neuronal ganglioside storage does not improve the clinical course of the Niemann-Pick C disease mouse. Hum Mol Genet. 2000;9:1087–1092.
    1. Gondre-Lewis MC, McGlynn R, Walkley SU. Cholesterol accumulation in NPC1-deficient neurons is ganglioside dependent. Curr Biol. 2003;13:1324–1329.
    1. Lachmann RH, Platt FM. Substrate reduction therapy for glycosphingolipid storage disorders. Expert Opin Investig Drugs. 2001;10:455–466.
    1. Zervas M, Somers KL, Thrall MA, Walkley SU. Critical role for glycosphingolipids in Niemann-Pick disease type C. Curr Biol. 2001;11:1283–1287.
    1. Patterson MC, Vecchio D, Prady H, Abel L, Wraith JE. Miglustat for treatment of Niemann-Pick C disease: a randomized controlled study. Lancet Neurol. 2007;6:765–772.
    1. Griffin LD, Gong W, Verot L, Mellon SH. Niemann-Pick type C disease involves disrupted neurosteriodogenesis and responds to allopregnanolone. Nat Med. 2004;10:704–711.
    1. Walkley SU. Pathogenic mechanisms in lysosomal disease: a reappraisal of the role of the lysosome. Acta Paediatr. 2007;(Suppl 96):26–32.
    1. Liu B, Li H, Repa JJ, Turley SD, Dietschy JM. Genetic variations and treatments that affect the lifespan of the NPC1 mouse. J Lipid Res. 2008;49:663–669.
    1. Liu B, Turley SD, Burns DK, Miller AM, Repa JJ, et al. Reversal of defective lysosomal transport in NPC disease ameliorates liver dysfunction and neurodegeneration in the npc1−/− mouse. PNAS USA. 2009;106:2377–2382.
    1. Pacheco CD, Kunkel R, Lieberman AP. Autophagy in Niemann-Pick C disease is dependent upon Beclin-1 and responsive to lipid trafficking defects. Hum Mol Genet. 2007;16:1495–1503.
    1. Liao G, Yao Y, Liu J, Yu Z, Cheung S, et al. Cholesterol accumulation is associated with lysosomal dysfunction and autophagic stress in Npc1−/− mouse brain. Am J Pathol. 2007;171:962–975.
    1. Jeyakumar M, Thomas R, Elliot-Smith E, Smith DA, van der Spoel AC, et al. Central nervous system inflammation is a hallmark of pathogenesis in mouse models of GM1 and GM2 gangliosidosis. Brain. 2003;126:974–987.
    1. Irie T, Uekama K. Pharmaceutical applications of cyclodextrins. III. Toxicological issues and safety evaluation. J Pharm Sci. 1997;86:147–162.
    1. Goldin E, Roff CF, Miller SPF, Rodriguez-Lafrasse C, Vanier MT, et al. Type C Niemann-Pick disease: a murine model of the lysosomal cholesterol lipidosis accumulates sphingosine and sphinganine in the liver. Biochim Biophys Acta. 1992;1127:303–311.
    1. Rodriguez-Lafrasse C, Rousson R, Pentchev PG, Louisot P, Vanier MT. Free sphingoid bases in tissues from patients with type C Niemann-Pick disease and other lysosomal storage disorders. Biochim Biophys Acta. 1994;1226:138–144.
    1. Lloyd-Evans E, Morgan AJ, He X, Smith DA, Elliot-Smith E, et al. Niemann-Pick disease type C1 is a sphingosine storage disease that causes deregulation of lysosomal calcium. Nat Med. 2008;14:1247–1255.
    1. Walkley SU. Secondary accumulation of gangliosides in lysosomal storage disorders. Semin Cell Dev Biol. 2004;15:433–444.
    1. Neufeld EF, Muenzer J. The mucopolysaccharidoses. In: Scriver CR, Beaudet AL, Sly WS, Valle D, editors. New York: McGraw-Hill Medical Publishing Division; 2001. pp. 3421–3452. The Metabolic and Molecular Basis of Inherited Disease, vol. 3, 8th edition.
    1. McGlynn R, Dobrenis K, Walkley SU. Differential subcellular localization of cholesterol, gangliosides, and glycosaminoglycans in murine models of mucopolysaccharide storage disorders. J Comp Neurol. 2004;480:415–426.
    1. Walkley SU, Vanier MT. Secondary lipid accumulation in lysosomal disease. Biochim Biophys Acta. 2009;1793:726–736.
    1. Hahn CN, del Pilar Martin M, Schroder M, Vanier MT, Hara Y, et al. Generalized CNS disease and massive GM1-ganglioside accumulation in mice defective in lysosomal acid β-galactosidase. Hum Mol Genet. 1997;6:205–211.
    1. Bhaumik M, Muller VJ, Rozaklis T, Johnson L, Dobrenis K, et al. A mouse model for mucopolysaccharidosis type III A (Sanfilippo syndrome). Glycobiology. 1999;9:1389–1396.
    1. Lau AA, Crawley AC, Hopwood JJ, Hemsley HM. Open field locomotor activity and anxiety-related behaviors in mucopolysaccharidosis type IIIA mice. Behav Brain Res. 2008;191:130–136.
    1. Camargo F, Erickson RP, Garver WS, Hossain GS, Carbone PN, et al. Cyclodextrins in the treatment of a mouse model of Niemann-Pick C disease. Life Sci. 2001;70:131–142.
    1. Monnaert V, Tilloy S, Bricout H, Fenart L, Cecchelli R, et al. Behavior of α-, β-, and γ-cyclodextrins and their derivatives on an in vitro model of blood-brain barrier. J Pharmacol Exp Ther. 2004;310:745–751.
    1. Jeulin H, Venard V, Carapito D, Finance C, Kedzierewicz F. Effective ribavirin concentration in mice brain using cyclodextrin as a drug carrier: Evaluation in a measles encephalitis model. Antiviral Res. 2009;81:261–266.
    1. Zidovetzki R, Levitan I. Use of cyclodextrins to manipulate plasma membrane cholesterol content: evidence, misconceptions and control strategies. Biochim Biophys Acta. 2007;1768:1311–1324.
    1. Chevallier J, Chamoun Z, Jiang G, Prestwich G, Sakai N, et al. Lysobisphosphatidic acid controls endosomal cholesterol levels. J Bio Chem. 2008;283:27871–27880.
    1. Deng D, Jiang N, Hao SJ, Sun H, Zhang GJ. Loss of membrane cholesterol influences lysosomal permeability to potassium ions and protons. Biochim Biophys Acta. 2009;1788:470–476.
    1. Lange Y, Ye J, Rigney M, Steck TL. Dynamics of lysosomal cholesterol in Niemann-Pick type C and normal human fibroblasts. J Lipid Res. 2002;43:198–204.
    1. Atger VM, de la Llera Moya M, Stoudt GW, Rodriguez WV, Phillips MC, et al. Cyclodextrins as catalysts for the removal of cholesterol from macrophage foam cells. J Clin Invest. 1997;99:773–780.
    1. Pfrieger FW. Outsourcing in the brain: do neurons depend on cholesterol delivery by astrocytes? Bioessays. 2003;25:72–78.
    1. Benarroch EE. Brain cholesterol metabolism and neurologic disease. Neurology. 2008;71:1368–1373.
    1. Zhang M, Strnatka D, Donohue C, Hallows JL, Vincent I, et al. Astrocyte-only Npc1 reduces neuronal cholesterol and triples lifespan of Npc1−/− mice. J Neurosci Res. 2008;86:2848–2856.
    1. Rajewski RA, Stella VJ. Pharmaceutical applications of cyclodextrins. 2. In vivo drug delivery. J Pharm Sci. 1996;85:1142–1169.
    1. Gould S, Scott RC. 2-Hydroxypropyl-beta-cyclodextrin (HP-beta-CD): a toxicology review. Food Chem Toxicol. 2005;43:1451–1459.
    1. Loftus SK, Morris JA, Carstea ED, Gu JZ, Cummings C, et al. Murine model of Niemann-Pick C disease: mutation in a cholesterol homeostasis gene. Science. 1997;277:232–235.
    1. Fujita N, Suzuki K, Vanier MT, Popko B, Maeda N, et al. Targeted disruption of the mouse sphingolipid activator protein gene: a complex phenotype, including severe leukodystrophy and wide-spread storage of multiple sphingolipids. Hum Mol Genet. 1996;5:711–725.
    1. Miscenyi MC, Dobrenis K, Stephney G, Pickel J, Vanier MT, et al. Neuropathology of the Mcoln1(−/−) knockout mouse model of mucolipidosis type IV. J Neuropathol Exp Neurol. 2009;68:125–135.

Source: PubMed

3
Sottoscrivi